1
|
Myburgh L, van Loon K, Huijbers EJM, van Beijnum JR, Russell CA, Griffioen AW. Guided design for the development of an evolution-proof influenza vaccine. Vaccine 2025; 59:127281. [PMID: 40398325 DOI: 10.1016/j.vaccine.2025.127281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/24/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Influenza remains a significant public health concern, particularly among high-risk populations, due to its capacity to cause annual epidemics and potentially trigger global pandemics. Despite the availability of countermeasures such as vaccines and antiviral treatments, their effectiveness is hindered by factors such as resistance development with manufacturing of the influenza vaccine still heavily relying on decades-old technologies. This review therefore examines the mechanisms by which influenza viruses evade host immunity and evaluates current and emerging approaches to enhance vaccine-mediated protection. Advances targeting the conserved hemagglutinin (HA) stem, incorporating multiple HA subtypes, and the use of adjuvants are discussed, alongside increased attention to neuraminidase (NA) and other viral components as immunogenic targets. Strategic epitope prediction, through glycan masking, evolutionary forecasting, and consensus sequence design, offer promising frameworks for rational vaccine design. Furthermore, delivery platforms, including recombinant protein, mRNA, and conjugate vaccines are explored for their potential to elicit broad and durable immunity. Collectively, these developments highlight a multifaceted approach towards the design of effective interventions against this persistent healthcare challenge.
Collapse
Affiliation(s)
- Lauren Myburgh
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Colin A Russell
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Amsterdam, UMC, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2025; 13:2164-2178. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
3
|
Larsen SE, Rais M, Reese VA, Ferede D, Pecor T, Kaur S, Nag D, Smytheman T, Gray SA, Carter D, Baldwin SL, Coler RN. Characterizing TLR4 agonist EmT4™ as an anti-Mycobacterium tuberculosis vaccine adjuvant. Immunohorizons 2025; 9:vlaf014. [PMID: 40285479 PMCID: PMC12032397 DOI: 10.1093/immhor/vlaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
Tuberculosis (TB) is again the deadliest infectious disease globally, and more efficacious vaccines are needed to reduce this mortality. Successful subunit TB vaccines need antigens and adjuvants that are immunogenic, inexpensive, and accessible. Here we evaluated the potential of synthetically produced Monophosphoryl lipid A (SyMLP), a TLR4-agonist, formulated in an oil-in-water emulsion (EmT4™) in combination with selected fusion proteins, to drive an effective vaccine-mediated immunogenic response in C57BL/6 mice against Mycobacterium tuberculosis (M.tb) HN878 and H37Rv challenge. We first observed that EmT4™ enhances activation of C57BL/6 bone-marrow derived macrophages and dendritic cells measured by CD40, CD86, and MHCII expression by flow cytometry. EmT4™ did not induce safety signals in a scaled tolerability study. In immunogenicity studies, mice immunized 3 times 3 weeks apart with ID93 antigen + EmT4™ produced a significantly higher magnitude of circulating proinflammatory cytokines and ID93-specific immunoglobulin G (IgG) antibodies pre- and post-challenge with M.tb than saline control animals. Ex vivo ID93 restimulated splenocytes and lung cells elicited significant polyfunctional CD4+ T-helper 1 responses. Importantly, ID93 + EmT4™ immunizations significantly reduced bacterial burden in C57BL/6 mice 4 weeks post-challenge. Interestingly, EmT4™ paired with a next generation protein fusion ID91 also afforded prophylactic protection against M.tb HN878 challenge in both young (6 to 8 wk) and aged (20 mo) immunocompromised Beige mice. These protection and immunogenicity findings suggest that synthetically derived EmT4™ adjuvant is not only suitable to help backfill the preclinical TB vaccine candidate pipeline but is also suitable for the needs of the global community.
Collapse
Affiliation(s)
- Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Valerie A Reese
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Debora Ferede
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Suhavi Kaur
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Deepika Nag
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Thomas Smytheman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Sean A Gray
- PAI Life Sciences Inc, Seattle, WA, United States
| | | | - Susan L Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
4
|
Tengattini S, Bavaro T, Rinaldi F, Temporini C, Pollegioni L, Terreni M, Piubelli L. Novel tuberculosis vaccines based on TB10.4 and Ag85B: State-of-art and advocacy for good practices. Vaccine 2025; 53:126932. [PMID: 40031085 DOI: 10.1016/j.vaccine.2025.126932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
Tuberculosis (TB) has plagued humanity in numerous devastating forms for centuries and remains a significant health challenge. Mycobacterium tuberculosis (Mtb), the bacterium responsible for TB, was the leading cause of death among infectious agents until the COVID-19 pandemic emerged. Immunization with the bacillus Calmette-Guérin (BCG) vaccine is one of the primary strategies to mitigate the risk of TB. Despite its widespread use, the current BCG vaccine has limited efficacy, particularly in adults. This review focuses on the rational design of vaccine candidates targeting the antigens TB10.4 and Ag85B. The review discusses the roles of TB10.4 and Ag85B in the virulence of Mtb and notes challenges in their production. Additionally, various protein conjugation strategies to enhance immunogenicity, including linking these antigens to glycans and adjuvants, are considered, as well as the most appropriate analytical methods for characterizing recombinant antigenic proteins and their conjugates. Finally, the associated challenges in developing a vaccine encompassing specific glycans and protein components were highlighted. We claim that using standardized procedures and detailed reporting in protein production and chemical modification can improve the reproducibility and rationalization of biological results. By adhering to these guidelines, the goal of developing an effective vaccine against TB will be best achieved.
Collapse
Affiliation(s)
- Sara Tengattini
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Teodora Bavaro
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Francesca Rinaldi
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Caterina Temporini
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
| | - Marco Terreni
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Luciano Piubelli
- Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy.
| |
Collapse
|
5
|
Nguyen NDNT, Subratheepam S, Guleed S, Melchiors KM, Olsen AW, Wørzner K, Follmann F, Dietrich J. Post-exposure vaccine protection of CTH522/CAF ®01 against reinfection with Chlamydia trachomatis requires Th1/Th17 but not Th2-immunity. NPJ Vaccines 2025; 10:65. [PMID: 40175404 PMCID: PMC11965518 DOI: 10.1038/s41541-025-01117-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/20/2025] [Indexed: 04/04/2025] Open
Abstract
Chlamydia trachomatis (C.t.) is globally the most common sexually transmitted bacterium with an estimated 131 million new cases occurring every year. There is no licenced vaccine against C.t. Repeated infections are often observed in women, suggesting that natural immunity is only partially protective. It is therefore important to investigate if a vaccine given post exposure, on top of a partially protective natural immunity, can increase protection against reinfection. In mice, an infection leads to robust immunity to subsequent challenges that precludes an investigation of increased protection elicited by a post-exposure vaccine. Therefore, we developed a new animal model where the first infection only provided partial protection against reinfection. Using this model, we show that UV-SvD/CAF®01 and CTH522/CAF®01 as post-exposure parenteral vaccines, but not CTH522/AlOH, protected against reinfection. As CTH522/CAF®01 also reduced the gross pathology score post reinfection, this suggests that CTH522/CAF®01 is both protective and safe as a post-exposure vaccine.
Collapse
Affiliation(s)
| | - Sharmila Subratheepam
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark
| | - Safia Guleed
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark
| | | | - Anja Weinreich Olsen
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark
| | - Katharina Wørzner
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark
| | - Frank Follmann
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark
| | - Jes Dietrich
- Statens Serum Institut, Department of Infectious Disease Immunology, Copenhagen, Denmark.
| |
Collapse
|
6
|
Khalifa AZ, Perrie Y, Shahiwala A. Subunit antigen delivery: emulsion and liposomal adjuvants for next-generation vaccines. Expert Opin Drug Deliv 2025; 22:583-597. [PMID: 40021342 DOI: 10.1080/17425247.2025.2474088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Developing new vaccines to combat emerging infectious diseases has gained more significance after the COVID-19 pandemic. Vaccination is the most cost-effective method for preventing infectious diseases, and subunit antigens are a safer alternative to traditional live, attenuated, and inactivated vaccines. AREAS COVERED Challenges in delivering subunit antigens and the status of different vaccine adjuvants. Recent research developments involving emulsion and liposomal adjuvants and their compositions and properties affecting their adjuvancy. EXPERT OPINION Lipid-based adjuvants, e.g. emulsions and liposomes, represent a paradigm shift in vaccine technology by enabling robust humoral and cellular immune responses with lower antigen doses, a property that is particularly critical during pandemics or in resource-limited settings. These adjuvants can optimize vaccine administration strategies by potentially reducing the frequency of booster doses, thereby improving patient compliance and lowering healthcare costs. While emulsions excel in dose-sparing and broadening immune responses, liposomes offer customization and precision in antigen delivery. However, the broader clinical application of these technologies is not without challenges. Stability issues, e.g. the susceptibility of emulsion-based adjuvants to freezing and their reliance on cold-chain logistics, pose significant barriers to their use in remote/underserved regions. Future developments will likely focus on improving manufacturing scalability and cost-effectiveness.
Collapse
Affiliation(s)
- Al Zahraa Khalifa
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Aliasgar Shahiwala
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
| |
Collapse
|
7
|
Beck CN, Santamaria JM, Erf GF. Inflammatory and Humoral Immune Responses to Commercial Autogenous Salmonella Bacterin Vaccines in Light-Brown Leghorn Pullets: Primary and Secondary Vaccine Responses. Vaccines (Basel) 2025; 13:311. [PMID: 40266196 PMCID: PMC11946197 DOI: 10.3390/vaccines13030311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Commercial poultry flocks undergo Salmonella vaccinations to manage salmonellosis outbreaks. Due to reports of severe injection site reactions to Salmonella bacterins, assessment of local inflammatory responses is necessary. The objective was to assess local inflammatory and systemic humoral immune responses to commercial autogenous Salmonella bacterin vaccines (SV1 or SV2) following primary or secondary intradermal (i.d.) vaccination in Light-Brown Leghorns (LBLs). METHODS LBL pullets received primary (14 wks) or secondary (19 wks) vaccination by i.d. growing feather (GF) pulp injection of SV1, SV2, Salmonella Enteritidis (SE) lipopolysaccharide (LPS), or water-oil-water emulsion (V). Local leukocyte levels and relative cytokine mRNA expression were monitored before (0 d) and at 6 h, 1 d, 2 d, 3 d, 5 d, and 7 d post-GF pulp injection (p.i.). Blood was collected through 28 d post-primary or -secondary vaccination, and SE-specific antibodies were quantified via ELISA. RESULTS Primary vaccine administration increased local heterophil and macrophage levels and increased IL-6 and IL-8 mRNA expressions at 6 h p.i., independent of treatment. Secondary administration extended these local immune activities through 3 d p.i. and included prolonged IL-17A mRNA expression. Primary and secondary GF-pulp injection with V resulted in rapid lymphocyte recruitment by 6 h p.i., comprised primarily of CD4+ and γδ T cells. SV1 and SV2 also produced a T-dependent systemic humoral immune response, as indicated by the IgM-to-IgG isotype switch, along with a memory phenotype in the secondary response. CONCLUSIONS These commercial-killed Salmonella vaccines, when prepared in water-oil-water emulsions, stimulated prolonged innate and T helper (Th) 17-type inflammatory responses at the injection site and produced a classic systemic humoral immune response after a second vaccination. Further research is needed to determine if extended inflammation influences adaptive immune responses in eliminating Salmonella infection.
Collapse
Affiliation(s)
- Chrysta N. Beck
- Department of Poultry Science, Division of Agriculture, University of Arkansas System, Fayetteville, AR 72701, USA;
| | | | - Gisela F. Erf
- Department of Poultry Science, Division of Agriculture, University of Arkansas System, Fayetteville, AR 72701, USA;
| |
Collapse
|
8
|
Islam EA, Fegan JE, Zeppa JJ, Ahn SK, Ng D, Currie EG, Lam J, Moraes TF, Gray-Owen SD. Adjuvant-dependent impacts on vaccine-induced humoral responses and protection in preclinical models of nasal and genital colonization by pathogenic Neisseria. Vaccine 2025; 48:126709. [PMID: 39817984 DOI: 10.1016/j.vaccine.2025.126709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/05/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
Neisseria gonorrhoeae, which causes the sexually transmitted infection gonorrhea and Neisseria meningitidis, a leading cause of bacterial meningitis and septicemia, are closely related human-restricted pathogens that inhabit distinct primary mucosal niches. While successful vaccines against invasive meningococcal disease have been available for decades, the rapid rise in antibiotic resistance has led to an urgent need to develop an effective gonococcal vaccine. Several surface antigens are shared among these two pathogens, making cross-species protection an exciting prospect. However, the type of vaccine-mediated immune response required to achieve protection against respiratory versus genital infection remains ill defined. In this study, we utilize well established mouse models of female lower genital tract colonization by N. gonorrhoeae and upper respiratory tract colonization by N. meningitidis to examine the performance of transferrin binding protein B (TbpB) vaccines formulated with immunologically distinct vaccine adjuvants. We demonstrate that vaccine-mediated protection is influenced by the choice of adjuvant, with Th1/2-balanced adjuvants performing optimally against N. gonorrhoeae, and both Th1/2-balanced and Th2-skewing adjuvants leading to a significant reduction in N. meningitidis burden. We further establish a lack of correlation between protection status and the humoral response or bactericidal titre. Combined, this work supports the feasibility for a single vaccine formulation to achieve pan-neisserial coverage.
Collapse
Affiliation(s)
- Epshita A Islam
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Jamie E Fegan
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Joseph J Zeppa
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Sang Kyun Ahn
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Dixon Ng
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Elissa G Currie
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Jessica Lam
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Trevor F Moraes
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Scott D Gray-Owen
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada.
| |
Collapse
|
9
|
Pellegrina D, Wilson HL, Mutwiri GK, Helmy M. Transcriptional Systems Vaccinology Approaches for Vaccine Adjuvant Profiling. Vaccines (Basel) 2025; 13:33. [PMID: 39852812 PMCID: PMC11768747 DOI: 10.3390/vaccines13010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Adjuvants are a diverse group of substances that can be added to vaccines to enhance antigen-specific immune responses and improve vaccine efficacy. The first adjuvants, discovered almost a century ago, were soluble crystals of aluminium salts. Over the following decades, oil emulsions, vesicles, oligodeoxynucleotides, viral capsids, and other complex organic structures have been shown to have adjuvant potential. However, the detailed mechanisms of how adjuvants enhance immune responses remain poorly understood and may be a barrier that reduces the rational selection of vaccine components. Previous studies on mechanisms of action of adjuvants have focused on how they activate innate immune responses, including the regulation of cell recruitment and activation, cytokine/chemokine production, and the regulation of some "immune" genes. This approach provides a narrow perspective on the complex events involved in how adjuvants modulate antigen-specific immune responses. A comprehensive and efficient way to investigate the molecular mechanism of action for adjuvants is to utilize systems biology approaches such as transcriptomics in so-called "systems vaccinology" analysis. While other molecular biology methods can verify if one or few genes are differentially regulated in response to vaccination, systems vaccinology provides a more comprehensive picture by simultaneously identifying the hundreds or thousands of genes that interact with complex networks in response to a vaccine. Transcriptomics tools such as RNA sequencing (RNA-Seq) allow us to simultaneously quantify the expression of practically all expressed genes, making it possible to make inferences that are only possible when considering the system as a whole. Here, we review some of the challenges in adjuvant studies, such as predicting adjuvant activity and toxicity when administered alone or in combination with antigens, or classifying adjuvants in groups with similar properties, while underscoring the significance of transcriptomics in systems vaccinology approaches to propel vaccine development forward.
Collapse
Affiliation(s)
- Diogo Pellegrina
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
| | - Heather L. Wilson
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - George K. Mutwiri
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Mohamed Helmy
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Computer Science, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
- Department of Computer Science, Idaho State University, Pocatello, ID 83209, USA
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| |
Collapse
|
10
|
Hojo-Souza NS, de Castro JT, Rivelli GG, Azevedo PO, Oliveira ER, Faustino LP, Salazar N, Bagno FF, Carvalho AF, Rattis B, Lourenço KL, Gomes IP, Assis BRD, Piccin M, Fonseca FG, Durigon E, Silva JS, de Souza RP, Goulart GAC, Santiago H, Fernandes APS, Teixeira SR, Gazzinelli RT. SpiN-Tec: A T cell-based recombinant vaccine that is safe, immunogenic, and shows high efficacy in experimental models challenged with SARS-CoV-2 variants of concern. Vaccine 2024; 42:126394. [PMID: 39368129 DOI: 10.1016/j.vaccine.2024.126394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 10/07/2024]
Abstract
The emergence of new SARS-CoV-2 variants of concern associated with waning immunity induced by natural infection or vaccines currently in use suggests that the COVID-19 pandemic will become endemic. Investing in new booster vaccines using different platforms is a promising way to enhance protection and keep the disease under control. Here, we evaluated the immunogenicity, efficacy, and safety of the SpiN-Tec vaccine, based on a chimeric recombinant protein (SpiN) adjuvanted with CTVad1 (MF59-based adjuvant), aiming at boosting immunity against variants of concern of SARS-CoV-2. Immunization of K18-hACE-2 transgenic mice and hamsters induced high antibody titers and cellular immune response to the SpiN protein as well as to its components, RBD and N proteins. Importantly in a heterologous prime/boost protocol with a COVID-19 vaccine approved for emergency use (ChAdOx1), SpiN-Tec enhanced the level of circulation neutralizing antibodies (nAb). In addition to protection against the Wuhan isolate, protection against the Delta and Omicron variants was also observed as shown by reduced viral load and lung pathology. Toxicity and safety tests performed in rats demonstrated that the SpiN-Tec vaccine was safe and, based on these results, the SpiN-Tec phase I/II clinical trial was approved.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Mice
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Cricetinae
- Mice, Transgenic
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- T-Lymphocytes/immunology
- Female
- Immunogenicity, Vaccine
- Humans
- Rats
- Disease Models, Animal
- Adjuvants, Vaccine
- Immunization, Secondary
- Vaccine Efficacy
Collapse
Affiliation(s)
- Natália S Hojo-Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | - Júlia T de Castro
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil; Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Graziella G Rivelli
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Patrick O Azevedo
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | | | - Lídia P Faustino
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil
| | - Natália Salazar
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Flávia F Bagno
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Alex F Carvalho
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Bruna Rattis
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Karine L Lourenço
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Isabela P Gomes
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil
| | - Bruna R D Assis
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Mariela Piccin
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Flávio G Fonseca
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Microbiologia, Universidade Federal de Minas Gerais, Brazil
| | - Edison Durigon
- Instituto de Ciências Biológicas, Universidade de São Paulo, Brazil
| | - João S Silva
- Plataforma Bi-Institucional de Pesquisa em Medicina Translacional, Fundação Oswaldo Cruz, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Renan P de Souza
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais, Brazil
| | - Gisele A C Goulart
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Helton Santiago
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil
| | - Ana Paula S Fernandes
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil
| | - Ricardo T Gazzinelli
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Brazil; Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Brazil; Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
11
|
Ontiveros-Padilla L, Bachelder EM, Ainslie KM. Microparticle and nanoparticle-based influenza vaccines. J Control Release 2024; 376:880-898. [PMID: 39427775 DOI: 10.1016/j.jconrel.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Influenza infections are a health public problem worldwide every year with the potential to become the next pandemic. Vaccination is the most effective strategy to prevent future influenza outbreaks, however, influenza vaccines need to be reformulated each year to provide protection due to viral antigenic drift and shift. As more efficient influenza vaccines are needed, it is relevant to recapitulate strategies to improve the immunogenicity and broad reactivity of the current vaccines. Here, we review the current approved vaccines in the U.S. market and the platform used for their production. We discuss the different approaches to develop a broadly reactive vaccine as well as reviewing the adjuvant systems that are under study for being potentially included in future influenza vaccine formulations. The main components of the immune system involved in achieving a protective immune response are reviewed and how they participate in the trafficking of particles systemically and in the mucosa. Finally, we describe and classify, according to their physicochemical properties, some of the potential micro and nano-particulate platforms that can be used as delivery systems for parenteral and mucosal vaccinations.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA; Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, UNC, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Weth AF, Dangerfield EM, Timmer MSM, Stocker BL. Recent Advances in the Development of Mincle-Targeting Vaccine Adjuvants. Vaccines (Basel) 2024; 12:1320. [PMID: 39771982 PMCID: PMC11680293 DOI: 10.3390/vaccines12121320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Abstract
The Macrophage-inducible C-type lectin (Mincle) is a pattern-recognition receptor (PRR), which has shown much promise as a molecular target for the development of TH1/TH17-skewing vaccine adjuvants. In 2009, the first non-proteinaceous Mincle ligands, trehalose dimycolate (TDM) and trehalose dibehenate (TDB), were identified. This prompted a search for other Mincle agonists and the exploration of Mincle agonists as vaccine adjuvants for both preventative and therapeutic (anti-cancer) vaccines. In this review, we discuss those classes of Mincle agonists that have been explored for their adjuvant potential. These Mincle agonists have been used as stand-alone adjuvants or in combination with other pathogen-associated molecular patterns (PAMPs) or immunomodulatory agents. We will also highlight recently identified Mincle ligands with hitherto unknown adjuvanticity. Conjugate vaccines that contain covalently linked adjuvants and/or adjuvant-antigen combinations are also presented, as well as the different formulations (e.g., oil-in-water emulsions, liposomes, and particulate delivery systems) that have been used for the codelivery of antigens and adjuvants. Insofar the reader is presented with a thorough review of the potential of Mincle-mediated vaccine adjuvants, including historical context, present-day research and clinical trials, and outstanding research questions, such as the role of ligand presentation and Mincle clustering, which, if better understood, will aid in the development of the much-needed TH1/TH17-skewing vaccine adjuvants.
Collapse
Affiliation(s)
| | | | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| |
Collapse
|
13
|
Woodworth JS, Contreras V, Christensen D, Naninck T, Kahlaoui N, Gallouët AS, Langlois S, Burban E, Joly C, Gros W, Dereuddre-Bosquet N, Morin J, Liu Olsen M, Rosenkrands I, Stein AK, Krøyer Wood G, Follmann F, Lindenstrøm T, Hu T, Le Grand R, Pedersen GK, Mortensen R. MINCLE and TLR9 agonists synergize to induce Th1/Th17 vaccine memory and mucosal recall in mice and non-human primates. Nat Commun 2024; 15:8959. [PMID: 39420177 PMCID: PMC11487054 DOI: 10.1038/s41467-024-52863-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Development of new vaccines tailored for difficult-to-target diseases is hampered by a lack of diverse adjuvants for human use, and none of the currently available adjuvants induce Th17 cells. Here, we develop a liposomal adjuvant, CAF®10b, that incorporates Mincle and Toll-like receptor 9 agonists. In parallel mouse and non-human primate studies comparing to CAF® adjuvants already in clinical trials, we report species-specific effects of adjuvant composition on the quality and magnitude of the responses. When combined with antigen, CAF®10b induces Th1 and Th17 responses and protection against a pulmonary infection with Mycobacterium tuberculosis in mice. In non-human primates, CAF®10b induces higher Th1 responses and robust Th17 responses detectable after six months, and systemic and pulmonary Th1 and Th17 recall responses, in a sterile model of local recall. Overall, CAF®10b drives robust memory antibody, Th1 and Th17 vaccine-responses via a non-mucosal immunization route across both rodent and primate species.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark.
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Sébastien Langlois
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Emma Burban
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Candie Joly
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Wesley Gros
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Julie Morin
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Ming Liu Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Ann-Kathrin Stein
- Department of Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Tu Hu
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark.
| |
Collapse
|
14
|
Hahn WO, Parks KR, Shen M, Ozorowski G, Janes H, Ballweber-Fleming L, Woodward Davis AS, Duplessis C, Tomai M, Dey AK, Sagawa ZK, De Rosa SC, Seese A, Kallur Siddaramaiah L, Stamatatos L, Lee WH, Sewall LM, Karlinsey D, Turner HL, Rubin V, Furth S, MacPhee K, Duff M, Corey L, Keefer MC, Edupuganti S, Frank I, Maenza J, Baden LR, Hyrien O, Sanders RW, Moore JP, Ward AB, Tomaras GD, Montefiori DC, Rouphael N, McElrath MJ. Use of 3M-052-AF with Alum adjuvant in HIV trimer vaccine induces human autologous neutralizing antibodies. J Exp Med 2024; 221:e20240604. [PMID: 39235529 PMCID: PMC11380150 DOI: 10.1084/jem.20240604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
Stabilized trimers preserving the native-like HIV envelope structure may be key components of a preventive HIV vaccine regimen to induce broadly neutralizing antibodies (bnAbs). We evaluated trimeric BG505 SOSIP.664 gp140 formulated with a novel TLR7/8 signaling adjuvant, 3M-052-AF/Alum, for safety, adjuvant dose-finding, and immunogenicity in a first-in-healthy adult (n = 17), randomized, and placebo-controlled trial (HVTN 137A). The vaccine regimen appeared safe. Robust, trimer-specific antibody, and B cell and CD4+ T cell responses emerged after vaccination. Five vaccinees developed serum autologous tier 2 nAbs (ID50 titer, 1:28-1:8647) after two to three doses targeting C3/V5 and/or V1/V2/V3 Env regions by electron microscopy and mutated pseudovirus-based neutralization analyses. Trimer-specific, B cell-derived monoclonal antibody activities confirmed these results and showed weak heterologous neutralization in the strongest responder. Our findings demonstrate the clinical utility of the 3M-052-AF/Alum adjuvant and support further improvements of trimer-based Env immunogens to focus responses on multiple broad nAb epitopes.
Collapse
Affiliation(s)
- William O. Hahn
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - K. Rachael Parks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mingchao Shen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | - Chris Duplessis
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | - Antu K. Dey
- International AIDS Vaccine Initiative, New York, NY, USA
| | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Leonidas Stamatatos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leigh M. Sewall
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Dalton Karlinsey
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Hannah L. Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vanessa Rubin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sarah Furth
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kellie MacPhee
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael Duff
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Srilatha Edupuganti
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ian Frank
- School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rogier W. Sanders
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Georgia D. Tomaras
- Center for Human Systems Immunology and Departments of Surgery and Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | | | | | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
15
|
Hoang-Phou S, Pal S, Slepenkin A, Abisoye-Ogunniyun A, Zhang Y, Gilmore SF, Shelby ML, Bourguet FA, Mohagheghi MV, Noy A, Rasley A, de la Maza LM, Coleman MA. CT584 Is Not a Protective Vaccine Antigen against Respiratory Chlamydial Challenge in Mice. Vaccines (Basel) 2024; 12:1134. [PMID: 39460301 PMCID: PMC11512284 DOI: 10.3390/vaccines12101134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background:Chlamydia trachomatis is the most prevalent bacterial sexually transmitted pathogen in humans worldwide. Since chlamydial infection is largely asymptomatic with the potential for serious complications, a preventative vaccine is likely the most viable long-term answer to this public health threat. Cell-free protein synthesis (CFPS) utilizes the cellular protein manufacturing machinery decoupled from the requirement for maintaining cellular viability, offering the potential for flexible, rapid, and decentralized production of recombinant protein vaccine antigens. Methods: Here, we use CFPS to produce the full-length putative chlamydial type three secretion system (T3SS) needle-tip protein, CT584, for evaluation as a vaccine antigen in mouse models. High-speed atomic force microscopy (HS-AFM) (RIBM, Tsukuba, Japan) imaging and computer simulations confirm that CFPS-produced CT584 retains a native-like structure prior to immunization. Female mice were primed with CT584 adjuvanted with CpG-1826 intranasally (i.n.) or CpG-1826 + Montanide ISA 720 intramuscularly (i.m.), followed four weeks later by an i.m. boost before respiratory challenge with 104 inclusion forming units (IFU) of Chlamydia muridarum. Results: Immunization with CT584 generated robust antibody responses but weak cell-mediated immunity and failed to protect against i.n. challenge as demonstrated by body weight loss, increased lung weights, and the presence of high numbers of IFUs in the lungs. Conclusion: While CT584 was not a protective vaccine candidate, the speed and flexibility with which CFPS can be used to produce other potential chlamydial antigens make it an attractive technique for antigen production.
Collapse
Affiliation(s)
- Steven Hoang-Phou
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA; (S.P.); (A.S.); (L.M.d.l.M.)
| | - Anatoli Slepenkin
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA; (S.P.); (A.S.); (L.M.d.l.M.)
| | - Abisola Abisoye-Ogunniyun
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Yuliang Zhang
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Sean F. Gilmore
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Megan L. Shelby
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Feliza A. Bourguet
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Mariam V. Mohagheghi
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Aleksandr Noy
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Amy Rasley
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| | - Luis M. de la Maza
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA; (S.P.); (A.S.); (L.M.d.l.M.)
| | - Matthew A. Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA; (S.H.-P.); (A.A.-O.); (Y.Z.); (S.F.G.); (M.L.S.); (F.A.B.); (M.V.M.); (A.N.); (A.R.)
| |
Collapse
|
16
|
Wang R, Oliveira LVN, Hester MM, Carlson D, Christensen D, Specht CA, Levitz SM. Protection against experimental cryptococcosis elicited by Cationic Adjuvant Formulation 01-adjuvanted subunit vaccines. PLoS Pathog 2024; 20:e1012220. [PMID: 38976694 PMCID: PMC11257399 DOI: 10.1371/journal.ppat.1012220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/18/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
The fungal infection, cryptococcosis, is responsible for >100,000 deaths annually. No licensed vaccines are available. We explored the efficacy and immune responses of subunit cryptococcal vaccines adjuvanted with Cationic Adjuvant Formulation 01 (CAF01). CAF01 promotes humoral and T helper (Th) 1 and Th17 immune responses and has been safely used in human vaccine trials. Four subcutaneous vaccines, each containing single recombinant Cryptococcus neoformans protein antigens, partially protected mice from experimental cryptococcosis. Protection increased, up to 100%, in mice that received bivalent and quadrivalent vaccine formulations. Vaccinated mice that received a pulmonary challenge with C. neoformans had an influx of leukocytes into the lung including robust numbers of polyfunctional CD4+ T cells which produced interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL)-17 upon ex vivo antigenic stimulation. Cytokine-producing lung CD8+ T cells were also found, albeit in lesser numbers. A significant, durable IFNγ response was observed in the lungs, spleen, and blood. Moreover, IFNγ secretion following ex vivo stimulation directly correlated with fungal control in the lungs. Thus, we have developed multivalent cryptococcal vaccines which protect mice from experimental cryptococcosis using an adjuvant which has been safely tested in humans. These preclinical studies suggest a path towards human cryptococcal vaccine trials.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Lorena V. N. Oliveira
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Maureen M. Hester
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Diana Carlson
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | | | - Charles A. Specht
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
17
|
Sanchez PL, Staats HF, Abraham SN, Ross TM. Mastoparan-7 adjuvanted COBRA H1 and H3 hemagglutinin influenza vaccines. Sci Rep 2024; 14:13800. [PMID: 38877101 PMCID: PMC11178843 DOI: 10.1038/s41598-024-64351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
Adjuvants enhance, prolong, and modulate immune responses by vaccine antigens to maximize protective immunity and enable more effective immunization in the young and elderly. Most adjuvants are formulated with injectable vaccines. However, an intranasal route of vaccination may induce mucosal and systemic immune responses for enhancing protective immunity in individuals and be easier to administer compared to injectable vaccines. In this study, a next generation of broadly-reactive influenza hemagglutinin (HA) vaccines were developed using the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology. These HA vaccines were formulated with Mastoparan 7 (M7-NH2) mast cell degranulating peptide adjuvant and administered intranasally to determine vaccine-induced seroconversion of antibodies against a panel of influenza viruses and protection following infection with H1N1 and H3N2 viruses in mice. Mice vaccinated intranasally with M7-NH2-adjuvanted COBRA HA vaccines had high HAIs against a panel of H1N1 and H3N2 influenza viruses and were protected against both morbidity and mortality, with reduced viral lung titers, following challenge with an H1N1 influenza virus. Additionally, M7-NH2 adjuvanted COBRA HA vaccines induced Th2 skewed immune responses with robust IgG and isotype antibodies in the serum and mucosal lung lavages. Overall, this intranasally delivered M7-NH2 -adjuvanted COBRA HA vaccine provides effective protection against drifted H1N1 and H3N2 viruses.
Collapse
Affiliation(s)
- Pedro L Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| | - Herman F Staats
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University, Duke University Medical Center, Durham, NC, USA
| | - Soman N Abraham
- Pathology Department, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, School of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA.
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
18
|
Hoang-Phou S, Pal S, Slepenkin A, Abisoye-Ogunniyun A, Zhang Y, Gilmore SF, Shelby M, Bourguet F, Mohagheghi M, Noy A, Rasley A, de la Maza LM, Coleman MA. Evaluation in mice of cell-free produced CT584 as a Chlamydia vaccine antigen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597210. [PMID: 38895407 PMCID: PMC11185655 DOI: 10.1101/2024.06.04.597210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Chlamydia trachomatis is the most prevalent bacterial sexually transmitted pathogen worldwide. Since chlamydial infection is largely asymptomatic with the potential for serious complications, a preventative vaccine is likely the most viable long-term answer to this public health threat. Cell-free protein synthesis (CFPS) utilizes the cellular protein manufacturing machinery decoupled from the requirement for maintaining cellular viability, offering the potential for flexible, rapid, and de-centralized production of recombinant protein vaccine antigens. Here, we use CFPS to produce the putative chlamydial type three secretion system (T3SS) needle-tip protein, CT584, for use as a vaccine antigen in mouse models. High-speed atomic force microscopy (HS-AFM) imaging and computer simulations confirm that CFPS-produced CT584 retains a native-like structure prior to immunization. Female mice were primed with CT584 adjuvanted with CpG-1826 intranasally (i.n.) or CpG-1826 + Montanide ISA 720 intramuscularly (i.m.), followed four-weeks later by an i.m. boost before respiratory challenge with 104 inclusion forming units (IFU) of Chlamydia muridarum. Immunization with CT584 generated robust antibody responses but weak cell mediated immunity and failed to protect against i.n. challenge as demonstrated by body weight loss, increased lungs' weights and the presence of high numbers of IFUs in the lungs. While CT584 alone may not be the ideal vaccine candidate, the speed and flexibility with which CFPS can be used to produce other potential chlamydial antigens makes it an attractive technique for antigen production.
Collapse
Affiliation(s)
- Steven Hoang-Phou
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Anatoli Slepenkin
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Abisola Abisoye-Ogunniyun
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Yuliang Zhang
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Sean F Gilmore
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Megan Shelby
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Feliza Bourguet
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Mariam Mohagheghi
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Aleksandr Noy
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Amy Rasley
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Matthew A Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94551, USA
| |
Collapse
|
19
|
Hahn WO, Parks KR, Shen M, Ozorowski G, Janes H, Ballweber-Fleming L, Woodward Davis AS, Duplessis C, Tomai M, Dey AK, Sagawa ZK, De Rosa SC, Seese A, Siddaramaiah LK, Stamatatos L, Lee WH, Sewall LM, Karlinsey D, Turner HL, Rubin V, Furth S, MacPhee K, Duff M, Corey L, Keefer MC, Edupuganti S, Frank I, Maenza J, Baden LR, Hyrien O, Sanders RW, Moore JP, Ward AB, Tomaras GD, Montefiori DC, Rouphael N, McElrath MJ. HIV BG505 SOSIP.664 trimer with 3M-052-AF/alum induces human autologous tier-2 neutralizing antibodies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.08.24306957. [PMID: 38766048 PMCID: PMC11100857 DOI: 10.1101/2024.05.08.24306957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Stabilized trimers preserving the native-like HIV envelope structure may be key components of a preventive HIV vaccine regimen to induce broadly neutralizing antibodies (bnAbs). We evaluated trimeric BG505 SOSIP.664 gp140, formulated with a novel TLR7/8 signaling adjuvant, 3M-052-AF/Alum, for safety, adjuvant dose-finding and immunogenicity in a first-in-healthy adult (n=17), randomized, placebo-controlled trial (HVTN 137A). The vaccine regimen appeared safe. Robust, trimer-specific antibody, B-cell and CD4+ T-cell responses emerged post-vaccination. Five vaccinees developed serum autologous tier-2 nAbs (ID50 titer, 1:28-1:8647) after 2-3 doses targeting C3/V5 and/or V1/V2/V3 Env regions by electron microscopy and mutated pseudovirus-based neutralization analyses. Trimer-specific, B-cell-derived monoclonal antibody activities confirmed these results and showed weak heterologous neutralization in the strongest responder. Our findings demonstrate the clinical utility of the 3M-052-AF/alum adjuvant and support further improvements of trimer-based Env immunogens to focus responses on multiple broad nAb epitopes. KEY TAKEAWAY/TAKE-HOME MESSAGES HIV BG505 SOSIP.664 trimer with novel 3M-052-AF/alum adjuvant in humans appears safe and induces serum neutralizing antibodies to matched clade A, tier 2 virus, that map to diverse Env epitopes with relatively high titers. The novel adjuvant may be an important mediator of vaccine response.
Collapse
|
20
|
Chan JYH, Clow F, Pearson V, Langley RJ, Fraser JD, Radcliff FJ. Feasibility of using a combination of staphylococcal superantigen-like proteins 3, 7 and 11 in a fusion vaccine for Staphylococcus aureus. Immunol Cell Biol 2024; 102:365-380. [PMID: 38572664 DOI: 10.1111/imcb.12745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
Staphylococcus aureus is a significant bacterial pathogen in both community and hospital settings, and the escalation of antimicrobial-resistant strains is of immense global concern. Vaccination is an inviting long-term strategy to curb staphylococcal disease, but identification of an effective vaccine has proved to be challenging. Three well-characterized, ubiquitous, secreted immune evasion factors from the staphylococcal superantigen-like (SSL) protein family were selected for the development of a vaccine. Wild-type SSL3, 7 and 11, which inhibit signaling through Toll-like receptor 2, cleavage of complement component 5 and neutrophil function, respectively, were successfully combined into a stable, active fusion protein (PolySSL7311). Vaccination of mice with an attenuated form of the PolySSL7311 protein stimulated significantly elevated specific immunoglobulin G and splenocyte proliferation responses to each component relative to adjuvant-only controls. Vaccination with PolySSL7311, but not a mixture of the individual proteins, led to a > 102 reduction in S. aureus tissue burden compared with controls after peritoneal challenge. Comparable antibody responses were elicited after coadministration of the vaccine in either AddaVax (an analog of MF59) or an Alum-based adjuvant; but only AddaVax conferred a significant reduction in bacterial load, aligning with other studies that suggest both cellular and humoral immune responses are necessary for protective immunity to S. aureus. Anti-sera from mice immunized with PolySSL7311, but not individual proteins, partially neutralized the functional activities of SSL7. This study confirms the importance of these SSLs for the survival of S. aureus in vivo and suggests that PolySSL7311 is a promising vaccine candidate.
Collapse
Affiliation(s)
- Janlin Ying Hui Chan
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Fiona Clow
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Victoria Pearson
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ries J Langley
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - John D Fraser
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Fiona J Radcliff
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
Wang R, Oliveira LVN, Hester MM, Carlson D, Christensen D, Specht CA, Levitz SM. Protection against experimental cryptococcosis elicited by Cationic Adjuvant Formulation 01-adjuvanted subunit vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.591045. [PMID: 38712080 PMCID: PMC11071535 DOI: 10.1101/2024.04.24.591045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The fungal infection, cryptococcosis, is responsible for >100,000 deaths annually. No licensed vaccines are available. We explored the efficacy and immune responses of subunit cryptococcal vaccines adjuvanted with Cationic Adjuvant Formulation 01 (CAF01). CAF01 promotes humoral and T helper (Th) 1 and Th17 immune responses and has been safely used in human vaccine trials. Four subcutaneous vaccines, each containing single recombinant Cryptococcus neoformans protein antigens, partially protected mice from experimental cryptococcosis. Protection increased, up to 100%, in mice that received bivalent and quadrivalent vaccine formulations. Vaccinated mice that received a pulmonary challenge with C. neoformans had an influx of leukocytes into the lung including robust numbers of polyfunctional CD4+ T cells which produced Interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL)-17 upon ex vivo antigenic stimulation. Cytokine-producing lung CD8+ T cells were also found, albeit in lesser numbers. A significant, durable IFNγ response was observed in the lungs, spleen, and blood. Moreover, IFNγ secretion following ex vivo stimulation directly correlated with fungal clearance in the lungs. Thus, we have developed multivalent cryptococcal vaccines which protect mice from experimental cryptococcosis using an adjuvant which has been safely tested in humans. These preclinical studies suggest a path towards human cryptococcal vaccine trials.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lorena V. N. Oliveira
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Maureen M. Hester
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Diana Carlson
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dennis Christensen
- Statens Serum Institut, Copenhagen, Denmark
- Present address: Croda Pharma, Diplomvej 381, Lyngby 2800, Denmark
| | - Charles A. Specht
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
22
|
Lavelle EC, McEntee CP. Vaccine adjuvants: Tailoring innate recognition to send the right message. Immunity 2024; 57:772-789. [PMID: 38599170 DOI: 10.1016/j.immuni.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Adjuvants play pivotal roles in vaccine development, enhancing immunization efficacy through prolonged retention and sustained release of antigen, lymph node targeting, and regulation of dendritic cell activation. Adjuvant-induced activation of innate immunity is achieved via diverse mechanisms: for example, adjuvants can serve as direct ligands for pathogen recognition receptors or as inducers of cell stress and death, leading to the release of immunostimulatory-damage-associated molecular patterns. Adjuvant systems increasingly stimulate multiple innate pathways to induce greater potency. Increased understanding of the principles dictating adjuvant-induced innate immunity will subsequently lead to programming specific types of adaptive immune responses. This tailored optimization is fundamental to next-generation vaccines capable of inducing robust and sustained adaptive immune memory across different cohorts.
Collapse
Affiliation(s)
- Ed C Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Craig P McEntee
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Wørzner K, Zimmermann J, Buhl R, Desoi A, Christensen D, Dietrich J, Nguyen NDNT, Lindenstrøm T, Woodworth JS, Alhakeem RS, Yu S, Ødum N, Mortensen R, Ashouri JF, Pedersen GK. Repeated immunization with ATRA-containing liposomal adjuvant transdifferentiates Th17 cells to a Tr1-like phenotype. J Autoimmun 2024; 144:103174. [PMID: 38377868 DOI: 10.1016/j.jaut.2024.103174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/17/2024] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
In many autoimmune diseases, autoantigen-specific Th17 cells play a pivotal role in disease pathogenesis. Th17 cells can transdifferentiate into other T cell subsets in inflammatory conditions, however, there have been no attempts to target Th17 cell plasticity using vaccines. We investigated if autoantigen-specific Th17 cells could be specifically targeted using a therapeutic vaccine approach, where antigen was formulated in all-trans retinoic acid (ATRA)-containing liposomes, permitting co-delivery of antigen and ATRA to the same target cell. Whilst ATRA was previously found to broadly reduce Th17 responses, we found that antigen formulated in ATRA-containing cationic liposomes only inhibited Th17 cells in an antigen-specific manner and not when combined with an irrelevant antigen. Furthermore, this approach shifted existing Th17 cells away from IL-17A expression and transcriptomic analysis of sorted Th17 lineage cells from IL-17 fate reporter mice revealed a shift of antigen-specific Th17 cells to exTh17 cells, expressing functional markers associated with T cell regulation and tolerance. In the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, vaccination with myelin-specific (MOG) antigen in ATRA-containing liposomes reduced Th17 responses and alleviated disease. This highlights the potential of therapeutic vaccination for changing the phenotype of existing Th17 cells in the context of immune mediated diseases.
Collapse
Affiliation(s)
- Katharina Wørzner
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.
| | - Julie Zimmermann
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Regitze Buhl
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anna Desoi
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Thomas Lindenstrøm
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Joshua S Woodworth
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Steven Yu
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, USA
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Denmark
| | - Rasmus Mortensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Judith F Ashouri
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, USA.
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Denmark
| |
Collapse
|
24
|
Tomás-Cortázar J, Quinn C, Corcoran N, Blanco A, Christensen D, McClean S. BpOmpW antigen administered with CAF01 adjuvant stimulates comparable T cell responses to Sigma adjuvant system. Vaccine X 2024; 17:100438. [PMID: 38303776 PMCID: PMC10831100 DOI: 10.1016/j.jvacx.2024.100438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
There are no licensed vaccines to protect vulnerable populations from the potentially fatal tropical infection, melioidosis, despite its causative agent, Burkholderia pseudomallei, being endemic in tropical and subtropical regions. A promising vaccine candidate, BpOmpW protected mice from melioidosis infection for up to 81 days and stimulated robust interferon gamma responses in CD4+, CD8+, NK and NKT cells. In order to progress to human studies, selection of an adjuvant with an acceptable human safety profile that stimulates appropriate correlates of protection is essential. Here we demonstrate that the CAF01 vaccine adjuvant elicits optimal immune correlates of protection when administered with our BpOmpW vaccine. Specifically, we demonstrate that CAF01 administered with BpOmpW elicits robust Th1 responses, with potent IFN-γ responses in CD4+ and CD8+ T cells and NKT cells, in addition to Th17 and Th2 responses. This formulation will be particularly effective in protecting susceptible populations including people with type 2 diabetes from melioidosis.
Collapse
Affiliation(s)
- Julen Tomás-Cortázar
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Conor Quinn
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Niamh Corcoran
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Alfonso Blanco
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen S, Denmark
| | - Siobhán McClean
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Hoseinpour R, Hasani A, Baradaran B, Abdolalizadeh J, Salehi R, Hasani A, Nabizadeh E, Yekani M, Hasani R, Kafil HS, Azizian K, Memar MY. Tuberculosis vaccine developments and efficient delivery systems: A comprehensive appraisal. Heliyon 2024; 10:e26193. [PMID: 38404880 PMCID: PMC10884459 DOI: 10.1016/j.heliyon.2024.e26193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Despite the widespread use of the Bacillus Calmette-Guérin (BCG) vaccine, Mycobacterium tuberculosis (MTB) continues to be a global burden. Vaccination has been proposed to prevent and treat tuberculosis (TB) infection, and several of them are in different phases of clinical trials. Though vaccine production is in progress but requires more attention. There are several TB vaccines in the trial phase, most of which are based on a combination of proteins/adjuvants or recombinant viral vectors used for selected MTB antigens. In this review, we attempted to discuss different types of TB vaccines based on the vaccine composition, the immune responses generated, and their clinical trial phases. Furthermore, we have briefly overviewed the effective delivery systems used for the TB vaccine and their effectiveness in different vaccines.
Collapse
Affiliation(s)
- Rasoul Hoseinpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Laboratory sciences and Microbiology, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Alka Hasani
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit, Sina Educational, Research, and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Applied Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Edris Nabizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Hossein Samadi Kafil
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Azizian
- Department of Microbiology, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Olsen AW, Rosenkrands I, Jacobsen CS, Cheeseman HM, Kristiansen MP, Dietrich J, Shattock RJ, Follmann F. Immune signature of Chlamydia vaccine CTH522/CAF®01 translates from mouse-to-human and induces durable protection in mice. Nat Commun 2024; 15:1665. [PMID: 38396019 PMCID: PMC10891140 DOI: 10.1038/s41467-024-45526-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/10/2024] [Indexed: 02/25/2024] Open
Abstract
The clinical development of an effective Chlamydia vaccine requires in-depth understanding of how well protective pre-clinical immune signatures translate to humans. Here, we report a comparative immunological characterization of CTH522/CAF®01 in female mice and humans. We find a range of immune signatures that translate from mouse to human, including a Th1/Th17 cytokine profile and antibody functionality. We identify vaccine-induced T cell epitopes, conserved among Chlamydia serovars, and previously found in infected individuals. Using the mouse model, we show that the common immune signature protected against ascending infection in mice, and vaccine induced antibodies could delay bacterial ascension to the oviduct, as well as development of pathology, in a T cell depleted mouse model. Finally, we demonstrate long-lasting immunity and protection of mice one year after vaccination. Based on the results obtained in the present study, we propose to further investigate CTH522/CAF®01 in a phase IIb study.
Collapse
Affiliation(s)
- Anja W Olsen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| | - Ida Rosenkrands
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Christina S Jacobsen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
- PharmaRelations, Virum, Denmark
| | | | - Max P Kristiansen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Frank Follmann
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
27
|
Hemmati S, Saeidikia Z, Seradj H, Mohagheghzadeh A. Immunomodulatory Peptides as Vaccine Adjuvants and Antimicrobial Agents. Pharmaceuticals (Basel) 2024; 17:201. [PMID: 38399416 PMCID: PMC10892805 DOI: 10.3390/ph17020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
The underdevelopment of adjuvant discovery and diversity, compared to core vaccine technology, is evident. On the other hand, antibiotic resistance is on the list of the top ten threats to global health. Immunomodulatory peptides that target a pathogen and modulate the immune system simultaneously are promising for the development of preventive and therapeutic molecules. Since investigating innate immunity in insects has led to prominent achievements in human immunology, such as toll-like receptor (TLR) discovery, we used the capacity of the immunomodulatory peptides of arthropods with concomitant antimicrobial or antitumor activity. An SVM-based machine learning classifier identified short immunomodulatory sequences encrypted in 643 antimicrobial peptides from 55 foe-to-friend arthropods. The critical features involved in efficacy and safety were calculated. Finally, 76 safe immunomodulators were identified. Then, molecular docking and simulation studies defined the target of the most optimal peptide ligands among all human cell-surface TLRs. SPalf2-453 from a crab is a cell-penetrating immunoadjuvant with antiviral properties. The peptide interacts with the TLR1/2 heterodimer. SBsib-711 from a blackfly is a TLR4/MD2 ligand used as a cancer vaccine immunoadjuvant. In addition, SBsib-711 binds CD47 and PD-L1 on tumor cells, which is applicable in cancer immunotherapy as a checkpoint inhibitor. MRh4-679 from a shrimp is a broad-spectrum or universal immunoadjuvant with a putative Th1/Th2-balanced response. We also implemented a pathway enrichment analysis to define fingerprints or immunological signatures for further in vitro and in vivo immunogenicity and reactogenicity measurements. Conclusively, combinatorial machine learning, molecular docking, and simulation studies, as well as systems biology, open a new opportunity for the discovery and development of multifunctional prophylactic and therapeutic lead peptides.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| | - Zahra Saeidikia
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| | - Hassan Seradj
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| | - Abdolali Mohagheghzadeh
- Department of Phytopharmaceuticals, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| |
Collapse
|
28
|
Poria R, Kala D, Nagraik R, Dhir Y, Dhir S, Singh B, Kaushik NK, Noorani MS, Kaushal A, Gupta S. Vaccine development: Current trends and technologies. Life Sci 2024; 336:122331. [PMID: 38070863 DOI: 10.1016/j.lfs.2023.122331] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Despite the effectiveness of vaccination in reducing or eradicating diseases caused by pathogens, there remain certain diseases and emerging infections for which developing effective vaccines is inherently challenging. Additionally, developing vaccines for individuals with compromised immune systems or underlying medical conditions presents significant difficulties. As well as traditional vaccine different methods such as inactivated or live attenuated vaccines, viral vector vaccines, and subunit vaccines, emerging non-viral vaccine technologies, including viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer new strategies to address the existing challenges in vaccine development. These advancements have also greatly enhanced our understanding of vaccine immunology, which will guide future vaccine development for a broad range of diseases, including rapidly emerging infectious diseases like COVID-19 and diseases that have historically proven resistant to vaccination. This review provides a comprehensive assessment of emerging non-viral vaccine production methods and their application in addressing the fundamental and current challenges in vaccine development.
Collapse
Affiliation(s)
- Renu Poria
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Deepak Kala
- Centera Laboratories, Institute of High Pressure Physics PAS, 01-142 Warsaw, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Yashika Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Bharat Singh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Naveen Kumar Kaushik
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| |
Collapse
|
29
|
Zimmermann J, van Haren SD, Diray-Arce J, Adriawan IR, Wørzner K, Krog RT, Guleed S, Hu T, Mortensen R, Dietrich J, Solbak SMØ, Levy O, Christensen D, Pedersen GK. Co-adjuvanting DDA/TDB liposomes with a TLR7 agonist allows for IgG2a/c class-switching in the absence of Th1 cells. NPJ Vaccines 2023; 8:189. [PMID: 38135685 PMCID: PMC10746746 DOI: 10.1038/s41541-023-00781-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Class-switching to IgG2a/c in mice is a hallmark response to intracellular pathogens. T cells can promote class-switching and the predominant pathway for induction of IgG2a/c antibody responses has been suggested to be via stimulation from Th1 cells. We previously formulated CAF®01 (cationic liposomes containing dimethyldioctadecylammonium bromide (DDA) and Trehalose-6,6-dibehenate (TDB)) with the lipidated TLR7/8 agonist 3M-052 (DDA/TDB/3M-052), which promoted robust Th1 immunity in newborn mice. When testing this adjuvant in adult mice using the recombinant Chlamydia trachomatis (C.t.) vaccine antigen CTH522, it similarly enhanced IgG2a/c responses compared to DDA/TDB, but surprisingly reduced the magnitude of the IFN-γ+Th1 response in a TLR7 agonist dose-dependent manner. Single-cell RNA-sequencing revealed that DDA/TDB/3M-052 liposomes initiated early transcription of class-switch regulating genes directly in pre-germinal center B cells. Mixed bone marrow chimeras further demonstrated that this adjuvant did not require Th1 cells for IgG2a/c switching, but rather facilitated TLR7-dependent T-bet programming directly in B cells. This study underlines that adjuvant-directed IgG2a/c class-switching in vivo can occur in the absence of T-cell help, via direct activation of TLR7 on B cells and positions DDA/TDB/3M-052 as a powerful adjuvant capable of eliciting type I-like immunity in B cells without strong induction of Th1 responses.
Collapse
Affiliation(s)
- Julie Zimmermann
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Katharina Wørzner
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ricki T Krog
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Safia Guleed
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Tu Hu
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Rasmus Mortensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Sara M Ø Solbak
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Christensen D. Head-to-Head Comparison of Novel Vaccine Technologies Comes with a Minefield of Challenges. Pharmaceutics 2023; 16:12. [PMID: 38276490 PMCID: PMC10819579 DOI: 10.3390/pharmaceutics16010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Modern vaccine development is having a golden period, with a variety of novel subunit technologies being introduced into clinical development in recent years. This opens the opportunity to find the best platform to use for novel vaccine antigen candidates through head-to-head comparative studies. Seldom appreciated is, however, the fact that these different technologies often do not have the same optimal antigen dose ratio, prime-boost regime and peak timepoint for measuring immunity. Instead, the preclinical studies that make the basis for platform selection use standard protocols not optimized for individual vaccines and fail to make selection on an informed basis. Here, I discuss the opportunities we have to optimize vaccine platform technologies through a better understanding of vaccine priming kinetics, the optimal antigen dose and sampling time and location.
Collapse
Affiliation(s)
- Dennis Christensen
- Adjuvant Systems Research & Development, Croda Pharma, 2800 Lyngby, Denmark
| |
Collapse
|
31
|
Okoth WA, Ho MF, Zaman M, Cooper E, Som P, Burgess M, Walton M, Nevagi RJ, Beattie L, Murphy D, Stanisic DI, Good MF. A CAF01-adjuvanted whole asexual blood-stage liposomal malaria vaccine induces a CD4 + T-cell-dependent strain-transcending protective immunity in rodent models. mBio 2023; 14:e0254723. [PMID: 37962347 PMCID: PMC10746282 DOI: 10.1128/mbio.02547-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/15/2023] Open
Abstract
IMPORTANCE Malaria is a devastating disease that has claimed many lives, especially children <5 years of age in Sub-Saharan Africa, as documented in World Malaria Reports by WHO. Even though vector control and chemoprevention tools have helped with elimination efforts in some, if not all, endemic areas, these efforts have been hampered by serious issues (including drug and insecticide resistance and disruption to social cohesion caused by the COVID-19 pandemic). Development of an effective malaria vaccine is the alternative preventative tool in the fight against malaria. Vaccines save millions of lives each year and have helped in elimination and/or eradication of global diseases. Development of a highly efficacious malaria vaccine that will ensure long-lasting protective immunity will be a "game-changing" prevention strategy to finally eradicate the disease. Such a vaccine will need to counteract the significant obstacles that have been hampering subunit vaccine development to date, including antigenic polymorphism, sub-optimal immunogenicity, and waning vaccine efficacy.
Collapse
Affiliation(s)
- Winter A. Okoth
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mei-Fong Ho
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Emily Cooper
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Priyanka Som
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mark Burgess
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Maddison Walton
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Reshma J. Nevagi
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Lynette Beattie
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Declan Murphy
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Michael F. Good
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
32
|
Hellfritzsch M, Christensen D, Foged C, Scherließ R, Thakur A. Reconstituted dry powder formulations of ZnO-adjuvanted ovalbumin induce equivalent antigen specific antibodies but lower T cell responses than ovalbumin adjuvanted with Alhydrogel® or cationic adjuvant formulation 01 (CAF®01). Int J Pharm 2023; 648:123581. [PMID: 37931728 DOI: 10.1016/j.ijpharm.2023.123581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Most licensed human vaccines are based on liquid dosage forms but have poor storage stability and require continuous and expensive cold-chain storage. In contrast, the use of solid vaccine dosage forms produced by for example spray drying, extends shelf life and eliminates the need for a cold chain. Zinc oxide (ZnO)-based nanoparticles display immunomodulatory properties, but their adjuvant effect as a dry powder formulation is unknown. Here, we show that reconstituted dry powder formulations of ZnO particles containing the model antigen ovalbumin (OVA) induce antigen-specific CD8+ T-cell and humoral responses. By systematically varying the ratio between ZnO and mannitol during spray drying, we manufactured dry powder formulations of OVA-containing ZnO particles that displayed: (i) a spherical or wrinkled surface morphology, (ii) an aerodynamic diameter and particle size distribution optimal for deep lung deposition, and (iii) aerosolization properties suitable for lung delivery. Reconstituted dry powder formulations of ZnO particles were well-tolerated by Calu-3 lung epithelial cells. Furthermore, almost equivalent OVA-specific serum antibody responses were stimulated by reconstituted ZnO particles, OVA adjuvanted with Alhydrogel®, and OVA adjuvanted with the cationic adjuvant formulation 01 (CAF®01). However, reconstituted dry powder ZnO particles and OVA adjuvanted with Alhydrogel® induced significantly lower OVA-specific CD8+CD44+ T-cell responses in the spleen than OVA adjuvanted with CAF®01. Similarly, reconstituted dry powder ZnO particles activated significantly lower percentages of follicular helper T cells and germinal center B cells in the draining lymph nodes than OVA adjuvanted with CAF®01. Overall, our results show that reconstituted dry powder formulations of ZnO nanoparticles can induce antigen-specific antibodies and can be used in vaccines to enhance antigen-specific humoral immune responses against subunit protein antigens.
Collapse
Affiliation(s)
- Marie Hellfritzsch
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany.
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
33
|
Chowdhury N, Kundu A. Nanotechnology Platform for Advancing Vaccine Development against the COVID-19 Virus. Diseases 2023; 11:177. [PMID: 38131983 PMCID: PMC10742622 DOI: 10.3390/diseases11040177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The COVID-19 pandemic has had a profound impact on societies, public health, healthcare systems, and the world economy. With over 771 million people infected worldwide and a staggering death toll exceeding 6,960,783 as of 4 October 2023 (according to the World Health Organization), the urgency for a solution was paramount. Since the outbreak, the demand for immediate treatment for COVID-19 viral infection, as well as for effective vaccination against this virus, was soaring, which led scientists, pharmaceutical/biotech companies, government health agencies, etc., to think about a treatment strategy that could control and minimize this outbreak as soon as possible. Vaccination emerged as the most effective strategy to combat this infectious disease. For vaccination strategies, any conventional vaccine approach using attenuated live or inactivated/engineered virus, as well as other approaches, typically requires years of research and assessment. However, the urgency of the situation promoted a faster and more effective approach to vaccine development against COVID-19. The role of nanotechnology in designing, manufacturing, boosting, and delivering vaccines to the host to counter this virus was unquestionably valued and assessed. Several nanoformulations are discussed here in terms of their composition, physical properties, credibility, and applications in past vaccine development (as well as the possibility of using those used in previous applications for the generation of the COVID-19 vaccine). Controlling and eliminating the spread of the virus and preventing future recurrence requires a safe, tolerable, and effective vaccine strategy. In this review, we discuss the potential of nanoformulations as the basis for an effective vaccine strategy against COVID-19.
Collapse
Affiliation(s)
| | - Anup Kundu
- Department of Biology, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| |
Collapse
|
34
|
Russi RC, del Balzo D, Reidel IG, Alonso Bivou M, Flor N, Lujan A, Sanchez D, Damiani MT, Veaute C. Evaluation of three formulations based on Polymorphic membrane protein D in mice infected with Chlamydia trachomatis. Front Immunol 2023; 14:1267684. [PMID: 38045697 PMCID: PMC10690417 DOI: 10.3389/fimmu.2023.1267684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
The significant impact of Chlamydia trachomatis(Ct) infections worldwide highlights the need to develop a prophylactic vaccine that elicits effective immunity and protects the host from the immunopathological effects of Ct infection. The aim of this study was to evaluate a vaccine based on a fragment of the Polymorphic membrane protein D (FPmpD) of C. trachomatis as an immunogen using a heterologous DNA prime-protein boost strategy in female mice Three different formulations were evaluated as protein boost: free recombinant FPmpD (rFPmpD) or rFPmpD formulated with a liposomal adjuvant alternatively supplemented with CpG or a cationic gemini lipopeptide as immunostimulants. The three candidates induced an increase in the cervicovaginal and systemic titers of anti-rFPmpD antibodies in two strains of mice (BALB/c and C57BL/6), with no evidence of fertility alterations. The three formulations induced a rapid and robust humoral immune response upon the Ct challenge. However, the booster with free rFPmpD more efficiently reduced the shedding of infective Ct and prevented the development of immunopathology. The formulations containing adjuvant induced a strong inflammatory reaction in the uterine tissue. Hence, the prime-boost strategy with the adjuvant-free FPmpD vaccine formulation might constitute a promissory candidate to prevent C. trachomatis intravaginal infection.
Collapse
Affiliation(s)
- Romina Cecilia Russi
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Diego del Balzo
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ivana Gabriela Reidel
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Mariano Alonso Bivou
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Noelia Flor
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Agustín Lujan
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Sanchez
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Teresa Damiani
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Carolina Veaute
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| |
Collapse
|
35
|
Thakur A, Wadhwa A, Lokras A, Müllertz OAO, Christensen D, Franzyk H, Foged C. Method of manufacturing CAF®09 liposomes affects immune responses induced by adjuvanted subunit proteins. Eur J Pharm Biopharm 2023; 189:84-97. [PMID: 37059402 DOI: 10.1016/j.ejpb.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
The ability to induce antigen-specific CD4+ and CD8+T-cell responses is one of the fundamental requirements when developing new efficacious vaccines against challenging infectious diseases and cancer. However, no adjuvants are currently approved for human subunit vaccines that induce T-cell immunity. Here, we incorporated a Toll-like receptor 4 agonist, i.e., the ionizable lipidoid L5N12, in the liposomal cationic adjuvant formulation 09 (CAF®09), and found that modified CAF®09 liposomes possess preserved adjuvant function as compared to unmodified CAF®09. CAF®09 consists of the cationic lipid dimethyldioctadecylammonium (DDA), monomycoloyl glycerol analogue 1 (MMG-1), and polyinosinic:polycytidylic acid [poly(I:C)]. By using the microfluidic mixing technology for liposome preparation, we gradually replaced DDA with L5N12, while keeping the molar ratios of MMG-1 and poly(I:C) constant. We found that this type of modification resulted in colloidally stable liposomes, which were significantly smaller and displayed reduced surface charge as compared to unmodified CAF®09, prepared by using the conventional thin film method. We showed that incorporation of L5N12 decreases the membrane rigidity of CAF®09 liposomes. Furthermore, vaccination with antigen adjuvanted with L5N12-modified CAF®09 or antigen adjuvanted with unmodified CAF®09, respectively, induced comparable antigen-specific serum antibody titers. We found that antigen adjuvanted with L5N12-modified CAF®09 induced antigen-specific effector and memory CD4+ and CD8+T-cell responses in the spleen comparable to those induced when unmodified CAF®09 was used as adjuvant. However, incorporating L5N12 did not have a synergistic immunopotentiating effect on the antibody and T-cell responses induced by CAF®09. Moreover, vaccination with antigen adjuvanted with unmodified CAF®09, which was manufactured by using microfluidic mixing, induced significantly lower antigen-specific CD4+ and CD8+T-cell responses than vaccination with antigen adjuvanted with unmodified CAF®09, which was prepared by using the thin film method. These results show that the method of manufacturing affects CAF®09 liposome adjuvanted antigen-specific immune responses, which should be taken into consideration when evaluating immunogenicity of subunit protein vaccines.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| | - Abishek Wadhwa
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Abhijeet Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
36
|
Nishiyama A, Adachi Y, Tonouchi K, Moriyama S, Sun L, Aoki M, Asanuma H, Shirakura M, Fukushima A, Yamamoto T, Takahashi Y. Post-fusion influenza vaccine adjuvanted with SA-2 confers heterologous protection via Th1-polarized, non-neutralizing antibody responses. Vaccine 2023; 41:4525-4533. [PMID: 37330368 DOI: 10.1016/j.vaccine.2023.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 06/19/2023]
Abstract
Development of a universal influenza vaccine that can provide robust and long-lasting protection against heterologous infections is a global public health priority. A variety of vaccine antigens are designed to increase the antigenicity of conserved epitopes to elicit cross-protective antibodies that often lack virus-neutralizing activity. Given the contribution of antibody effector functions to cross-protection, adjuvants need to be added to modulate antibody effector functions as well as to enhance antibody quantity. We previously showed that post-fusion influenza vaccine antigens elicit non-neutralizing but cross-protective antibodies against conserved epitopes. Here, using a murine model, we comparably assessed the adjuvanticity of the newly developed SA-2 adjuvant containing a synthetic TLR7 agonist DSP-0546 and squalene-based MF59 analog as representative Th1- or Th2-type adjuvants, respectively. Both types of adjuvants in the post-fusion vaccine comparably enhanced cross-reactive IgG titers against heterologous strains. However, only SA-2 skewed the IgG subclass into the IgG2c subclass in association to its Th1-polarizing nature. SA-2-enhanced IgG2c responses exhibited antibody-dependent cellular cytotoxicity against heterologous virus strains, without cross-neutralizing activity. Eventually, the SA-2-adjuvanted vaccination provided protection against lethal infection by heterologous H3N2 and H1N1 viruses. Together, we conclude that the combination with a SA-2 is advantageous for enhancing the cross-protective capability of post-fusion HA vaccines that elicit non-neutralizing IgG antibodies.
Collapse
Affiliation(s)
- Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan; Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan
| | - Keisuke Tonouchi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan; Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsucho Shinjuku, Tokyo 162-8480, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan
| | - Lin Sun
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan
| | - Masamitsu Aoki
- Sumitomo Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Hideki Asanuma
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo 208-0011, Japan
| | - Masayuki Shirakura
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo 208-0011, Japan
| | - Akihisa Fukushima
- Sumitomo Pharma. Co., Ltd., 3-1-98, Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan; Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan.
| |
Collapse
|
37
|
Corripio-Miyar Y, MacLeod CL, Mair I, Mellanby RJ, Moore BD, McNeilly TN. Self-Adjuvanting Calcium-Phosphate-Coated Microcrystal-Based Vaccines Induce Pyroptosis in Human and Livestock Immune Cells. Vaccines (Basel) 2023; 11:1229. [PMID: 37515044 PMCID: PMC10385459 DOI: 10.3390/vaccines11071229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Successful vaccines require adjuvants able to activate the innate immune system, eliciting antigen-specific immune responses and B-cell-mediated antibody production. However, unwanted secondary effects and the lack of effectiveness of traditional adjuvants has prompted investigation into novel adjuvants in recent years. Protein-coated microcrystals modified with calcium phosphate (CaP-PCMCs) in which vaccine antigens are co-immobilised within amino acid crystals represent one of these promising self-adjuvanting vaccine delivery systems. CaP-PCMCs has been shown to enhance antigen-specific IgG responses in mouse models; however, the exact mechanism of action of these microcrystals is currently unclear. Here, we set out to investigate this mechanism by studying the interaction between CaP-PCMCs and mammalian immune cells in an in vitro system. Incubation of cells with CaP-PCMCs induced rapid pyroptosis of peripheral blood mononuclear cells and monocyte-derived dendritic cells from cattle, sheep and humans, which was accompanied by the release of interleukin-1β and the activation of Caspase-1. We show that this pyroptotic event was cell-CaP-PCMCs contact dependent, and neither soluble calcium nor microcrystals without CaP (soluble PCMCs) induced pyroptosis. Our results corroborate CaP-PCMCs as a promising delivery system for vaccine antigens, showing great potential for subunit vaccines where the enhancement or find tuning of adaptive immunity is required.
Collapse
Affiliation(s)
| | - Clair Lyle MacLeod
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XQ, UK
| | - Iris Mair
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Richard J Mellanby
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, UK
| | - Barry D Moore
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XQ, UK
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Penicuik EH26 0PZ, UK
| |
Collapse
|
38
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
39
|
Dijkman K, Lindenstrøm T, Rosenkrands I, Søe R, Woodworth JS, Lindestam Arlehamn CS, Mortensen R. A protective, single-visit TB vaccination regimen by co-administration of a subunit vaccine with BCG. NPJ Vaccines 2023; 8:66. [PMID: 37160970 PMCID: PMC10169149 DOI: 10.1038/s41541-023-00666-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
The only licensed tuberculosis (TB) vaccine, Bacillus Calmette Guerin (BCG), fails to reliably protect adolescents and adults from pulmonary TB, resulting in ~1.6 million deaths annually. Protein subunit vaccines have shown promise against TB in clinical studies. Unfortunately, most subunit vaccines require multiple administrations, which increases the risk of loss to follow-up and necessitates more complex and costly logistics. Given the well-documented adjuvant effect of BCG, we hypothesized that BCG co-administration could compensate for a reduced number of subunit vaccinations. To explore this, we developed an expression-optimized version of our H107 vaccine candidate (H107e), which does not cross-react with BCG. In the CAF®01 adjuvant, a single dose of H107e induced inferior protection compared to three H107e/CAF®01 administrations. However, co-administering a single dose of H107e/CAF®01 with BCG significantly improved protection, which was equal to BCG co-administered with three H107e/CAF®01 doses. Importantly, combining BCG with a single H107e/CAF®01 dose also increased protection in previously BCG-primed animals. Overall, a single dose of H107e/CAF®01 with BCG induced long-lived immunity and triggered BCG-specific Th17 responses. These data support co-administration of BCG and subunit vaccines in both BCG naïve and BCG-primed individuals as an improved TB vaccine strategy with reduced number of vaccination visits.
Collapse
Affiliation(s)
- Karin Dijkman
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Rikke Søe
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| |
Collapse
|
40
|
Volosnikova EA, Merkuleva IA, Esina TI, Shcherbakov DN, Borgoyakova MB, Isaeva AA, Nesmeyanova VS, Volkova NV, Belenkaya SV, Zaykovskaya AV, Pyankov OV, Starostina EV, Zadorozhny AM, Zaitsev BN, Karpenko LI, Ilyichev AA, Danilenko ED. SARS-CoV-2 RBD Conjugated to Polyglucin, Spermidine, and dsRNA Elicits a Strong Immune Response in Mice. Vaccines (Basel) 2023; 11:vaccines11040808. [PMID: 37112720 PMCID: PMC10146165 DOI: 10.3390/vaccines11040808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the rapid development and approval of several COVID vaccines based on the full-length spike protein, there is a need for safe, potent, and high-volume vaccines. Considering the predominance of the production of neutralizing antibodies targeting the receptor-binding domain (RBD) of S-protein after natural infection or vaccination, it makes sense to choose RBD as a vaccine immunogen. However, due to its small size, RBD exhibits relatively poor immunogenicity. Searching for novel adjuvants for RBD-based vaccine formulations is considered a good strategy for enhancing its immunogenicity. Herein, we assess the immunogenicity of severe acute respiratory syndrome coronavirus 2 RBD conjugated to a polyglucin:spermidine complex (PGS) and dsRNA (RBD-PGS + dsRNA) in a mouse model. BALB/c mice were immunized intramuscularly twice, with a 2-week interval, with 50 µg of RBD, RBD with Al(OH)3, or conjugated RBD. A comparative analysis of serum RBD-specific IgG and neutralizing antibody titers showed that PGS, PGS + dsRNA, and Al(OH)3 enhanced the specific humoral response in animals. There was no significant difference between the groups immunized with RBD-PGS + dsRNA and RBD with Al(OH)3. Additionally, the study of the T-cell response in animals showed that, unlike adjuvants, the RBD-PGS + dsRNA conjugate stimulates the production of specific CD4+ and CD8+ T cells in animals.
Collapse
Affiliation(s)
- Ekaterina A Volosnikova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Iuliia A Merkuleva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Tatiana I Esina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Dmitry N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Mariya B Borgoyakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anastasiya A Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Valentina S Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Natalia V Volkova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Svetlana V Belenkaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Anna V Zaykovskaya
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Oleg V Pyankov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Ekaterina V Starostina
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexey M Zadorozhny
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Boris N Zaitsev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Larisa I Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Alexander A Ilyichev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| | - Elena D Danilenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Koltsovo, Russia
| |
Collapse
|
41
|
Borghi M, Gallinaro A, Pirillo MF, Canitano A, Michelini Z, De Angelis ML, Cecchetti S, Tinari A, Falce C, Mariotti S, Capocefalo A, Chiantore MV, Iacobino A, Di Virgilio A, van Gils MJ, Sanders RW, Lo Presti A, Nisini R, Negri D, Cara A. Different configurations of SARS-CoV-2 spike protein delivered by integrase-defective lentiviral vectors induce persistent functional immune responses, characterized by distinct immunogenicity profiles. Front Immunol 2023; 14:1147953. [PMID: 37090707 PMCID: PMC10113491 DOI: 10.3389/fimmu.2023.1147953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/13/2023] [Indexed: 04/08/2023] Open
Abstract
Several COVID-19 vaccine strategies utilizing new formulations for the induction of neutralizing antibodies (nAbs) and T cell immunity are still under evaluation in preclinical and clinical studies. Here we used Simian Immunodeficiency Virus (SIV)-based integrase defective lentiviral vector (IDLV) delivering different conformations of membrane-tethered Spike protein in the mouse immunogenicity model, with the aim of inducing persistent nAbs against multiple SARS-CoV-2 variants of concern (VoC). Spike modifications included prefusion-stabilizing double proline (2P) substitutions, mutations at the furin cleavage site (FCS), D614G mutation and truncation of the cytoplasmic tail (delta21) of ancestral and Beta (B.1.351) Spike, the latter mutation to markedly improve IDLV membrane-tethering. BALB/c mice were injected once with IDLV delivering the different forms of Spike or the recombinant trimeric Spike protein with 2P substitutions and FCS mutations in association with a squalene-based adjuvant. Anti-receptor binding domain (RBD) binding Abs, nAbs and T cell responses were detected up to six months from a single immunization with escalating doses of vaccines in all mice, but with different levels and kinetics. Results indicated that IDLV delivering the Spike protein with all the combined modifications, outperformed the other candidates in terms of T cell immunity and level of both binding Abs and nAbs soon after the single immunization and persistence over time, showing the best capacity to neutralize all formerly circulating VoC Alpha, Beta, Gamma and Delta. Although present, the lowest response was detected against Omicron variants (BA.1, BA.2 and BA.4/5), suggesting that the magnitude of immune evasion may be related to the higher genetic distance of Omicron as indicated by increased number of amino acid substitutions in Spike acquired during virus evolution.
Collapse
Affiliation(s)
- Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Serena Cecchetti
- Confocal Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Falce
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Sabrina Mariotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio Capocefalo
- Department of Veterinary Public Health & Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | | | - Angelo Iacobino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio Di Virgilio
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | | | - Roberto Nisini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Donatella Negri, ; Andrea Cara,
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Donatella Negri, ; Andrea Cara,
| |
Collapse
|
42
|
Vono M, Mastelic-Gavillet B, Mohr E, Östensson M, Persson J, Olafsdottir TA, Lemeille S, Pejoski D, Hartley O, Christensen D, Andersen P, Didierlaurent AM, Harandi AM, Lambert PH, Siegrist CA. C-type lectin receptor agonists elicit functional IL21-expressing Tfh cells and induce primary B cell responses in neonates. Front Immunol 2023; 14:1155200. [PMID: 37063899 PMCID: PMC10102809 DOI: 10.3389/fimmu.2023.1155200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionC-type lectin receptor (CLR) agonists emerged as superior inducers of primary B cell responses in early life compared with Toll-like receptor (TLR) agonists, while both types of adjuvants are potent in adults.MethodsHere, we explored the mechanisms accounting for the differences in neonatal adjuvanticity between a CLR-based (CAF®01) and a TLR4-based (GLA-SE) adjuvant administered with influenza hemagglutinin (HA) in neonatal mice, by using transcriptomics and systems biology analyses.ResultsOn day 7 after immunization, HA/CAF01 increased IL6 and IL21 levels in the draining lymph nodes, while HA/GLA-SE increased IL10. CAF01 induced mixed Th1/Th17 neonatal responses while T cell responses induced by GLA-SE had a more pronounced Th2-profile. Only CAF01 induced T follicular helper (Tfh) cells expressing high levels of IL21 similar to levels induced in adult mice, which is essential for germinal center (GC) formation. Accordingly, only CAF01- induced neonatal Tfh cells activated adoptively transferred hen egg lysozyme (HEL)-specific B cells to form HEL+ GC B cells in neonatal mice upon vaccination with HEL-OVA.DiscussionCollectively, the data show that CLR-based adjuvants are promising neonatal and infant adjuvants due to their ability to harness Tfh responses in early life.
Collapse
Affiliation(s)
- Maria Vono
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- *Correspondence: Maria Vono,
| | - Beatris Mastelic-Gavillet
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Elodie Mohr
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Malin Östensson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | | | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - David Pejoski
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Oliver Hartley
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Arnaud M. Didierlaurent
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ali M. Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, British Columbia (BC) Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Paul-Henri Lambert
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
43
|
Woodworth JS, Contreras V, Christensen D, Naninck T, Kahlaoui N, Gallouët AS, Langlois S, Burban E, Joly C, Gros W, Dereuddre-Bosquet N, Morin J, Olsen ML, Rosenkrands I, Stein AK, Wood GK, Follmann F, Lindenstrøm T, LeGrand R, Pedersen GK, Mortensen R. A novel adjuvant formulation induces robust Th1/Th17 memory and mucosal recall responses in Non-Human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529651. [PMID: 36865310 PMCID: PMC9980079 DOI: 10.1101/2023.02.23.529651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
After clean drinking water, vaccination is the most impactful global health intervention. However, development of new vaccines against difficult-to-target diseases is hampered by the lack of diverse adjuvants for human use. Of particular interest, none of the currently available adjuvants induce Th17 cells. Here, we develop and test an improved liposomal adjuvant, termed CAF®10b, that incorporates a TLR-9 agonist. In a head-to-head study in non-human primates (NHPs), immunization with antigen adjuvanted with CAF®10b induced significantly increased antibody and cellular immune responses compared to previous CAF® adjuvants, already in clinical trials. This was not seen in the mouse model, demonstrating that adjuvant effects can be highly species specific. Importantly, intramuscular immunization of NHPs with CAF®10b induced robust Th17 responses that were observed in circulation half a year after vaccination. Furthermore, subsequent instillation of unadjuvanted antigen into the skin and lungs of these memory animals led to significant recall responses including transient local lung inflammation observed by Positron Emission Tomography-Computed Tomography (PET-CT), elevated antibody titers, and expanded systemic and local Th1 and Th17 responses, including >20% antigen-specific T cells in the bronchoalveolar lavage. Overall, CAF®10b demonstrated an adjuvant able to drive true memory antibody, Th1 and Th17 vaccine-responses across rodent and primate species, supporting its translational potential.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Sébastien Langlois
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Emma Burban
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Candie Joly
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Wesley Gros
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Julie Morin
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Ming Liu Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Ann-Kathrin Stein
- Department of Vaccine Development, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Roger LeGrand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| |
Collapse
|
44
|
Müllertz OAO, Andersen P, Christensen D, Foged C, Thakur A. Pulmonary Administration of the Liposome-Based Adjuvant CAF01: Effect of Surface Charge on Mucosal Adjuvant Function. Mol Pharm 2023; 20:953-970. [PMID: 36583936 DOI: 10.1021/acs.molpharmaceut.2c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mucosal surfaces of the lungs represent a major site of entry for airborne pathogens, and pulmonary administration of vaccines is an attractive strategy to induce protective mucosal immunity in the airways. Recently, we demonstrated the potential of pulmonary vaccination with the tuberculosis subunit antigen H56 adjuvanted with the cationic liposomal adjuvant formulation CAF01, which consists of the cationic lipid dimethyldioctadecylammonium (DDA) bromide and the synthetic cord factor trehalose-6,6'-dibehenate. However, the cationic charge of DDA represents a major safety challenge. Hence, replacing DDA with a safer zwitterionic or anionic phospholipid is an attractive approach to improve vaccine safety, but the effect of liposomal surface charge on the induction of mucosal immunity after airway immunization is poorly understood. Here, we investigated the effect of surface charge by replacing the cationic DDA component of CAF01 with zwitterionic dipalmitoylphosphatidylcholine (DPPC) or anionic dipalmitoylphosphatidylglycerol (DPPG), and we show that charge modification enhances antigen-specific pulmonary T-cell responses against co-formulated H56. We systematically replaced DDA with either DPPC or DPPG and found that these modifications resulted in colloidally stable liposomes that have similar size and morphology to unmodified CAF01. DPPC- or DPPG-modified CAF01 displayed surface charge-dependent protein adsorption and induced slightly higher follicular helper T cells and germinal center B cells in the lung-draining lymph nodes than unmodified CAF01. In addition, modified CAF01 induced significantly higher levels of H56-specific Th17 cells and polyfunctional CD4+ T cells in the lungs, as compared to unmodified CAF01. However, the strong H56-specific humoral responses induced by CAF01 in the lungs and spleen were not influenced by surface charge. Hence, these results provide insights into the importance of surface charge for liposomal adjuvant function and can also guide the design of safe pulmonary subunit vaccines against other mucosal pathogens.
Collapse
Affiliation(s)
- Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| |
Collapse
|
45
|
Recombinant Protein Vaccines Formulated with Enantio-Specific Cationic Lipid R-DOTAP Induce Protective Cellular and Antibody-Mediated Immune Responses in Mice. Viruses 2023; 15:v15020432. [PMID: 36851646 PMCID: PMC9965888 DOI: 10.3390/v15020432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Adjuvants are essential components of subunit vaccines added to enhance immune responses to antigens through immunomodulation. Very few adjuvants have been approved for human use by regulatory agencies due to safety concerns. Current subunit vaccine adjuvants approved for human use are very effective in promoting humoral immune responses but are less effective at promoting T-cell immunity. In this study, we evaluated a novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP) as an immunomodulator for subunit vaccines capable of inducing both humoral- and cellular-mediated immunity. Using recombinant protein antigens derived from SARS-CoV2 spike or novel computationally optimized broadly reactive influenza antigen (COBRA) proteins, we demonstrated that R-DOTAP nanoparticles promoted strong cellular- and antibody-mediated immune responses in both monovalent and bivalent vaccines. R-DOTAP-based vaccines induced antigen-specific and polyfunctional CD8+ and CD4+ effector T cells and memory T cells, respectively. Antibody responses induced by R-DOTAP showed a balanced Th1/Th2 type immunity, neutralizing activity and protection of mice from challenge with live SARS-CoV2 or influenza viruses. R-DOTAP also facilitated significant dose sparing of the vaccine antigens. These studies demonstrate that R-DOTAP is an excellent immune stimulator for the production of next-generation subunit vaccines containing multiple recombinant proteins.
Collapse
|
46
|
Carnet F, Perrin-Cocon L, Paillot R, Lotteau V, Pronost S, Vidalain PO. An inventory of adjuvants used for vaccination in horses: the past, the present and the future. Vet Res 2023; 54:18. [PMID: 36864517 PMCID: PMC9983233 DOI: 10.1186/s13567-023-01151-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/27/2023] [Indexed: 03/04/2023] Open
Abstract
Vaccination is one of the most widely used strategies to protect horses against pathogens. However, available equine vaccines often have limitations, as they do not always provide effective, long-term protection and booster injections are often required. In addition, research efforts are needed to develop effective vaccines against emerging equine pathogens. In this review, we provide an inventory of approved adjuvants for equine vaccines worldwide, and discuss their composition and mode of action when available. A wide range of adjuvants are used in marketed vaccines for horses, the main families being aluminium salts, emulsions, polymers, saponins and ISCOMs. We also present veterinary adjuvants that are already used for vaccination in other species and are currently evaluated in horses to improve equine vaccination and to meet the expected level of protection against pathogens in the equine industry. Finally, we discuss new adjuvants such as liposomes, polylactic acid polymers, inulin, poly-ε-caprolactone nanoparticles and co-polymers that are in development. Our objective is to help professionals in the horse industry understand the composition of marketed equine vaccines in a context of mistrust towards vaccines. Besides, this review provides researchers with a list of adjuvants, either approved or at least evaluated in horses, that could be used either alone or in combination to develop new vaccines.
Collapse
Affiliation(s)
- Flora Carnet
- grid.508204.bLABÉO, 14280 Saint-Contest, France ,grid.412043.00000 0001 2186 4076BIOTARGEN, Normandie University, UNICAEN, 14280 Saint-Contest, France
| | - Laure Perrin-Cocon
- grid.462394.e0000 0004 0450 6033CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007 Lyon, France
| | - Romain Paillot
- grid.451003.30000 0004 0387 5232School of Equine and Veterinary Physiotherapy, Writtle University College, Lordship Road, Writtle, Chelmsford, CM1 3RR UK
| | - Vincent Lotteau
- grid.462394.e0000 0004 0450 6033CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007 Lyon, France
| | - Stéphane Pronost
- LABÉO, 14280, Saint-Contest, France. .,BIOTARGEN, Normandie University, UNICAEN, 14280, Saint-Contest, France.
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
47
|
DnaJ, a promising vaccine candidate against Ureaplasma urealyticum infection. Appl Microbiol Biotechnol 2022; 106:7643-7659. [PMID: 36269329 PMCID: PMC9589543 DOI: 10.1007/s00253-022-12230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/20/2022] [Accepted: 10/05/2022] [Indexed: 11/08/2022]
Abstract
Abstract
Ureaplasma urealyticum (U. urealyticum, Uu) is a common sexually transmitted pathogen that is responsible for diseases such as non-gonococcal urethritis, chorioamnionitis, and neonatal respiratory diseases. The rapid emergence of multidrug-resistant bacteria threatens the effective treatment of Uu infections. Considering this, vaccination could be an efficacious medical intervention to prevent Uu infection and disease. As a highly conserved molecular chaperone, DnaJ is expressed and upregulated by pathogens soon after infection. Here, we assessed the vaccine potential of recombinant Uu-DnaJ in a mouse model and dendritic cells. Results showed that intramuscular administration of DnaJ induced robust humoral- and T helper (Th) 1 cell-mediated immune responses and protected against genital tract infection, inflammation, and the pathologic sequelae after Uu infection. Importantly, the DnaJ protein also induced the maturation of mouse bone marrow–derived dendritic cells (BMDCs), ultimately promoting naïve T cell differentiation toward the Th1 phenotype. In addition, adoptive immunization of DnaJ-pulsed BMDCs elicited antigen-specific Immunoglobulin G2 (IgG2) antibodies as well as a Th1-biased cellular response in mice. These results support DnaJ as a promising vaccine candidate to control Uu infections. Key points • A novel recombinant vaccine was constructed against U. urealyticum infection. • Antigen-specific humoral and cellular immune responses after DnaJ vaccination. • Dendritic cells are activated by Uu-DnaJ, which results in a Th1-biased immune response. Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12230-4.
Collapse
|
48
|
Choque-Guevara R, Poma-Acevedo A, Montesinos-Millán R, Rios-Matos D, Gutiérrez-Manchay K, Montalvan-Avalos A, Quiñones-Garcia S, Cauti-Mendoza MDG, Agurto-Arteaga A, Ramirez-Ortiz I, Criollo-Orozco M, Huaccachi-Gonzales E, Romero YK, Perez-Martinez N, Isasi-Rivas G, Sernaque-Aguilar Y, Villanueva-Pérez D, Ygnacio F, Vallejos-Sánchez K, Fernández-Sánchez M, Guevara-Sarmiento LA, Fernández-Díaz M, Zimic M, for the COVID-19 Working Group in Perú. Squalene in oil-based adjuvant improves the immunogenicity of SARS-CoV-2 RBD and confirms safety in animal models. PLoS One 2022; 17:e0269823. [PMID: 35998134 PMCID: PMC9397949 DOI: 10.1371/journal.pone.0269823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/30/2022] [Indexed: 01/09/2023] Open
Abstract
COVID-19 pandemic has accelerated the development of vaccines against its etiologic agent, SARS-CoV-2. However, the emergence of new variants of the virus lead to the generation of new alternatives to improve the current sub-unit vaccines in development. In the present report, the immunogenicity of the Spike RBD of SARS-CoV-2 formulated with an oil-in-water emulsion and a water-in-oil emulsion with squalene was evaluated in mice and hamsters. The RBD protein was expressed in insect cells and purified by chromatography until >95% purity. The protein was shown to have the appropriate folding as determined by ELISA and flow cytometry binding assays to its receptor, as well as by its detection by hamster immune anti-S1 sera under non-reducing conditions. In immunization assays, although the cellular immune response elicited by both adjuvants were similar, the formulation based in water-in-oil emulsion and squalene generated an earlier humoral response as determined by ELISA. Similarly, this formulation was able to stimulate neutralizing antibodies in hamsters. The vaccine candidate was shown to be safe, as demonstrated by the histopathological analysis in lungs, liver and kidney. These results have shown the potential of this formulation vaccine to be evaluated in a challenge against SARS-CoV-2 and determine its ability to confer protection.
Collapse
Affiliation(s)
| | | | | | - Dora Rios-Matos
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Stefany Quiñones-Garcia
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maria de Grecia Cauti-Mendoza
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | | | - Yomara K. Romero
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Gisela Isasi-Rivas
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Freddy Ygnacio
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | - Katherine Vallejos-Sánchez
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | - Mirko Zimic
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
- * E-mail:
| | | |
Collapse
|
49
|
Hassanzadeh P, Atyabi F, Dinarvand R. Nanobionics: From plant empowering to the infectious disease treatment. J Control Release 2022; 349:890-901. [PMID: 35901860 DOI: 10.1016/j.jconrel.2022.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Infectious diseases (ID) are serious threats against the global health and socio-economic conditions. Vaccination usually plays a key role in disease prevention, however, insufficient efficiency or immunogenicity may be quite challenging. Using the advanced vectors for delivery of vaccines with suitable efficiency, safety, and immune-modulatory activity, and tunable characteristics could be helpful, but there are no systematic reviews confirming the capabilities of the vaccine delivery systems for covering various types of pathogens. Furthermore, high rates of the infections, transmission, and fatal ratio and diversity of the pathogens and infection mechanisms may negatively influence vaccine effectiveness. The absence of highly-effective antibiotics against the resistant strains of bacteria and longevity of antibiotic testing have provoked increasing needs towards the application of more accurate and specific theranostic strategies including the nanotechnology-based ones. Nanobionics which is based on the charge storage and transport in the molecular structures, could be of key value in the molecular diagnostic tests and highly-specific electro-analytical methods or devices. Such devices based on the early disease diagnostics might be of critical significance against various types of diseases. This article highlights the significance of nanobionics against ID.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran; Sasan Hospital, Tehran 14159-83391, Iran.
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| |
Collapse
|
50
|
Aradottir Pind AA, Thorsdottir S, Magnusdottir G, Meinke A, Del Giudice G, Jonsdottir I, Bjarnarson SP. A comparative study of adjuvants effects on neonatal plasma cell survival niche in bone marrow and persistence of humoral immune responses. Front Immunol 2022; 13:904415. [PMID: 35990686 PMCID: PMC9381929 DOI: 10.3389/fimmu.2022.904415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
The neonatal immune system is distinct from the immune system of older individuals rendering neonates vulnerable to infections and poor responders to vaccination. Adjuvants can be used as tools to enhance immune responses to co-administered antigens. Antibody (Ab) persistence is mediated by long-lived plasma cells that reside in specialized survival niches in the bone marrow, and transient Ab responses in early life have been associated with decreased survival of plasma cells, possibly due to lack of survival factors. Various cells can secrete these factors and which cells are the main producers is still up for debate, especially in early life where this has not been fully addressed. The receptor BCMA and its ligand APRIL have been shown to be important in the maintenance of plasma cells and Abs. Herein, we assessed age-dependent maturation of a broad range of bone marrow accessory cells and their expression of the survival factors APRIL and IL-6. Furthermore, we performed a comparative analysis of the potential of 5 different adjuvants; LT-K63, mmCT, MF59, IC31 and alum, to enhance expression of survival factors and BCMA following immunization of neonatal mice with tetanus toxoid (TT) vaccine. We found that APRIL expression was reduced in the bone marrow of young mice whereas IL-6 expression was higher. Eosinophils, macrophages, megakaryocytes, monocytes and lymphocytes were important secretors of survival factors in early life but undefined cells also constituted a large fraction of secretors. Immunization and adjuvants enhanced APRIL expression but decreased IL-6 expression in bone marrow cells early after immunization. Furthermore, neonatal immunization with adjuvants enhanced the proportion of plasmablasts and plasma cells that expressed BCMA both in spleen and bone marrow. Enhanced BCMA expression correlated with enhanced vaccine-specific humoral responses, even though the effect of alum on BCMA was less pronounced than those of the other adjuvants at later time points. We propose that low APRIL expression in bone marrow as well as low BCMA expression of plasmablasts/plasma cells in early life together cause transient Ab responses and could represent targets to be triggered by vaccine adjuvants to induce persistent humoral immune responses in this age group.
Collapse
Affiliation(s)
- Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sigrun Thorsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Gudbjorg Julia Magnusdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- *Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|